Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters











Publication year range
1.
Hum Mol Genet ; 31(16): 2738-2750, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35348691

ABSTRACT

Mutations in the X-linked cyclin-dependent kinase-like 5 (CDKL5) cause CDKL5 deficiency disorder (CDD), a neurodevelopmental disease characterized by severe infantile seizures and intellectual disability. The absence of CDKL5 in mice causes defective spine maturation that can at least partially explain the cognitive impairment in CDKL5 patients and CDD mouse models. The molecular basis for such defect may depend on the capacity of CDKL5 to regulate microtubule (MT) dynamics through its association with the MT-plus end tracking protein CLIP170 (cytoplasmic linker protein 170). Indeed, we here demonstrate that the absence of CDKL5 causes CLIP170 to be mainly in a closed inactive conformation that impedes its binding to MTs. Previously, the synthetic pregnenolone analogue, pregnenolone-methyl-ether (PME), was found to have a positive effect on CDKL5-related cellular and neuronal defects in vitro. Here, we show that PME induces the open active conformation of CLIP170 and promotes the entry of MTs into dendritic spines in vitro. Furthermore, the administration of PME to symptomatic Cdkl5-knock-out mice improved hippocampal-dependent behavior and restored spine maturation and the localization of MT-related proteins in the synaptic compartment. The positive effect on cognitive deficits persisted for 1 week after treatment withdrawal. Altogether, our results suggest that CDKL5 regulates spine maturation and cognitive processes through its control of CLIP170 and MT dynamics, which may represent a novel target for the development of disease-modifying therapies.


Subject(s)
Epileptic Syndromes , Microtubule-Associated Proteins , Neoplasm Proteins , Pregnenolone , Animals , Epileptic Syndromes/genetics , Ethers/metabolism , Hippocampus/metabolism , Mice , Microtubule-Associated Proteins/genetics , Microtubules/metabolism , Neoplasm Proteins/genetics , Pregnenolone/pharmacology , Protein Serine-Threonine Kinases/genetics
2.
J Neuroendocrinol ; 34(2): e13033, 2022 02.
Article in English | MEDLINE | ID: mdl-34495563

ABSTRACT

Pregnenolone methyl-ether (PME) is a synthetic derivative of the endogenous neuroactive steroid pregnenolone (PREG), which is an important modulator of several brain functions. In addition to being the precursor of steroids, PREG acts directly on various targets including microtubules (MTs), the functioning of which is fundamental for the development and homeostasis of nervous system. The coordination of MT dynamics is supported by a plethora of MT-associated proteins (MAPs) and by a specific MT code that is defined by the post-translational modifications of tubulin. Defects associated with MAPs or tubulin post-translational modifications are linked to different neurological pathologies including mood and neurodevelopmental disorders. In this review, we describe the beneficial effect of PME in major depressive disorders (MDDs) and in CDKL5 deficiency disorder (CDD), two pathologies that are joint by defective MT dynamics. Growing evidence indeed suggests that PME, as well as PREG, is able to positively affect the MT-binding of MAP2 and the plus-end tracking protein CLIP170 that are both found to be deregulated in the above mentioned pathologies. Furthermore, PME influences the state of MT acetylation, the deregulation of which is often associated with neurological abnormalities including MDDs. By contrast to PREG, PME is not metabolised into other downstream molecules with specific biological properties, an aspect that makes this compound more suitable for therapeutic strategies. Thus, through the analysis of MDDs and CDD, this work focuses attention on the possible use of PME for neuronal pathologies associated with MT defects.


Subject(s)
Depressive Disorder, Major , Methyl Ethers , Epileptic Syndromes , Humans , Methyl Ethers/metabolism , Methyl Ethers/pharmacology , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/pharmacology , Microtubules/metabolism , Pregnenolone/metabolism , Pregnenolone/therapeutic use , Protein Serine-Threonine Kinases , Spasms, Infantile , Tubulin/metabolism , Tubulin/pharmacology
3.
Int J Mol Sci ; 24(1)2022 Dec 21.
Article in English | MEDLINE | ID: mdl-36613509

ABSTRACT

CDKL5 deficiency disorder (CDD) is an X-linked neurodevelopmental disorder characterised by early-onset drug-resistant epilepsy and impaired cognitive and motor skills. CDD is caused by mutations in cyclin-dependent kinase-like 5 (CDKL5), which plays a well-known role in regulating excitatory neurotransmission, while its effect on neuronal inhibition has been poorly investigated. We explored the potential role of CDKL5 in the inhibitory compartment in Cdkl5-KO male mice and primary hippocampal neurons and found that CDKL5 interacts with gephyrin and collybistin, two crucial organisers of the inhibitory postsynaptic sites. Through molecular and electrophysiological approaches, we demonstrated that CDKL5 loss causes a reduced number of gephyrin puncta and surface exposed γ2 subunit-containing GABAA receptors, impacting the frequency of miniature inhibitory postsynaptic currents, which we ascribe to a postsynaptic function of CDKL5. In line with previous data showing that CDKL5 loss impacts microtubule (MT) dynamics, we showed that treatment with pregnenolone-methyl-ether (PME), which promotes MT dynamics, rescues the above defects. The impact of CDKL5 deficiency on inhibitory neurotransmission might explain the presence of drug-resistant epilepsy and cognitive defects in CDD patients. Moreover, our results may pave the way for drug-based therapies that could bypass the need for CDKL5 and provide effective therapeutic strategies for CDD patients.


Subject(s)
Neurosteroids , Spasms, Infantile , Male , Mice , Animals , Neurosteroids/therapeutic use , Pregnenolone/pharmacology , Spasms, Infantile/genetics , Ethers , Mice, Knockout , Protein Serine-Threonine Kinases/genetics
5.
EMBO Mol Med ; 12(6): e10270, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32383329

ABSTRACT

Mutations in MECP2 cause several neurological disorders of which Rett syndrome (RTT) represents the best-defined condition. Although mainly working as a transcriptional repressor, MeCP2 is a multifunctional protein revealing several activities, the involvement of which in RTT remains obscure. Besides being mainly localized in the nucleus, MeCP2 associates with the centrosome, an organelle from which primary cilia originate. Primary cilia function as "sensory antennae" protruding from most cells, and a link between primary cilia and mental illness has recently been reported. We herein demonstrate that MeCP2 deficiency affects ciliogenesis in cultured cells, including neurons and RTT fibroblasts, and in the mouse brain. Consequently, the cilium-related Sonic Hedgehog pathway, which is essential for brain development and functioning, is impaired. Microtubule instability participates in these phenotypes that can be rescued by HDAC6 inhibition together with the recovery of RTT-related neuronal defects. Our data indicate defects of primary cilium as a novel pathogenic mechanism that by contributing to the clinical features of RTT might impact on proper cerebellum/brain development and functioning, thus providing a novel therapeutic target.


Subject(s)
Rett Syndrome , Animals , Brain/metabolism , Hedgehog Proteins , Humans , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mice , Mutation , Rett Syndrome/genetics
6.
Int J Mol Sci ; 20(17)2019 Aug 21.
Article in English | MEDLINE | ID: mdl-31438497

ABSTRACT

CDKL5 deficiency disorder (CDD) is a severe neurodevelopmental encephalopathy caused by mutations in the X-linked CDKL5 gene that encodes a serine/threonine kinase. CDD is characterised by the early onset of seizures and impaired cognitive and motor skills. Loss of CDKL5 in vitro and in vivo affects neuronal morphology at early and late stages of maturation, suggesting a link between CDKL5 and the neuronal cytoskeleton. Recently, various microtubule (MT)-binding proteins have been identified as interactors of CDKL5, indicating that its roles converge on regulating MT functioning. MTs are dynamic structures that are important for neuronal morphology, migration and polarity. The delicate control of MT dynamics is fundamental for proper neuronal functions, as evidenced by the fact that aberrant MT dynamics are involved in various neurological disorders. In this review, we highlight the link between CDKL5 and MTs, discussing how CDKL5 deficiency may lead to deranged neuronal functions through aberrant MT dynamics. Finally, we discuss whether the regulation of MT dynamics through microtubule-targeting agents may represent a novel strategy for future pharmacological approaches in the CDD field.


Subject(s)
Epileptic Syndromes/metabolism , Microtubules/metabolism , Neurons/metabolism , Spasms, Infantile/metabolism , Animals , Humans , Microtubules/drug effects , Neurons/drug effects , Pregnenolone/pharmacology
7.
J Psychopharmacol ; 33(7): 894-907, 2019 07.
Article in English | MEDLINE | ID: mdl-31084246

ABSTRACT

BACKGROUND: Recent evidence suggests that 2-week treatment with the non-psychotomimetic cannabinoid cannabidivarin (CBDV) could be beneficial towards neurological and social deficits in early symptomatic Mecp2 mutant mice, a model of Rett syndrome (RTT). AIM: The aim of this study was to provide further insights into the efficacy of CBDV in Mecp2-null mice using a lifelong treatment schedule (from 4 to 9 weeks of age) to evaluate its effect on recognition memory and neurological defects in both early and advanced stages of the phenotype progression. METHODS: CBDV 0.2, 2, 20 and 200 mg/kg/day was administered to Mecp2-null mice from 4 to 9 weeks of age. Cognitive and neurological defects were monitored during the whole treatment schedule. Biochemical analyses were carried out in brain lysates from 9-week-old wild-type and knockout mice to evaluate brain-derived neurotrophic factor (BDNF) and insulin-like growth factor-1 (IGF-1) levels as well as components of the endocannabinoid system. RESULTS: CBDV rescues recognition memory deficits in Mecp2 mutant mice and delays the appearance of neurological defects. At the biochemical level, it normalizes BDNF/IGF1 levels and the defective PI3K/AKT/mTOR pathway in Mecp2 mutant mice at an advanced stage of the disease. Mecp2 deletion upregulates CB1 and CB2 receptor levels in the brain and these changes are restored after CBDV treatment. CONCLUSIONS: CBDV administration exerts an enduring rescue of memory deficits in Mecp2 mutant mice, an effect that is associated with the normalization of BDNF, IGF-1 and rpS6 phosphorylation levels as well as CB1 and CB2 receptor expression. CBDV delays neurological defects but this effect is only transient.


Subject(s)
Cannabinoids/pharmacology , Cognitive Dysfunction/drug therapy , Memory Disorders/drug therapy , Methyl-CpG-Binding Protein 2/genetics , Animals , Brain/drug effects , Brain/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Cannabinoids/administration & dosage , Cognitive Dysfunction/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mice, Knockout , Rett Syndrome/drug therapy , Rett Syndrome/physiopathology , Ribosomal Protein S6/metabolism
8.
Mol Neurobiol ; 56(7): 4838-4854, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30402709

ABSTRACT

MeCP2 is a fundamental protein associated with several neurological disorders, including Rett syndrome. It is considered a multifunctional factor with a prominent role in regulating chromatin structure; however, a full comprehension of the consequences of its deficiency is still lacking. Here, we characterize a novel mouse model of Mecp2 bearing the human mutation Y120D, which is localized in the methyl-binding domain. As most models of Mecp2, the Mecp2Y120D mouse develops a severe Rett-like phenotype. This mutation alters the interaction of the protein with chromatin, but surprisingly, it also impairs its association with corepressors independently on the involved interacting domains. These features, which become overt mainly in the mature brain, cause a more accessible and transcriptionally active chromatin structure; conversely, in the Mecp2-null brain, we find a less accessible and transcriptionally inactive chromatin. By demonstrating that different MECP2 mutations can produce concordant neurological phenotypes but discordant molecular features, we highlight the importance of considering personalized approaches for the treatment of Rett syndrome.


Subject(s)
Behavior, Animal , Gene Knock-In Techniques , Methyl-CpG-Binding Protein 2/metabolism , Precision Medicine , Animals , Brain/metabolism , Brain/pathology , Chromatin/metabolism , Female , Humans , Longevity , Male , Memory, Short-Term , Mice , Mice, Inbred C57BL , Models, Biological , Mutation/genetics , Neurons/metabolism , Phenotype , Rett Syndrome
9.
Neuropharmacology ; 144: 104-114, 2019 01.
Article in English | MEDLINE | ID: mdl-30326240

ABSTRACT

Mutations in the X-linked cyclin-dependent kinase-like 5 (CDKL5) gene cause CDKL5 Deficiency Disorder (CDD), a rare neurodevelopmental syndrome characterized by severe behavioural and physiological symptoms. No cure is available for CDD. CDKL5 is a kinase that is abundantly expressed in the brain and plays a critical role in neurodevelopmental processes, such as neuronal morphogenesis and plasticity. This study provides the first characterization of the neurobehavioural phenotype of 1 year old Cdkl5-null mice and demonstrates that stimulation of the serotonin receptor 7 (5-HT7R) with the agonist molecule LP-211 (0.25 mg/kg once/day for 7 days) partially rescues the abnormal phenotype and brain molecular alterations in Cdkl5-null male mice. In particular, LP-211 treatment completely normalizes the prepulse inhibition defects observed in Cdkl5-null mice and, at a molecular level, restores the abnormal cortical phosphorylation of rpS6, a downstream target of mTOR and S6 kinase, which plays a direct role in regulating protein synthesis. Moreover, we demonstrate for the first time that mitochondria show prominent functional abnormalities in Cdkl5-null mouse brains that can be restored by pharmacological stimulation of brain 5-HT7R.


Subject(s)
Brain/drug effects , Epileptic Syndromes/drug therapy , Mitochondria/drug effects , Piperazines/pharmacology , Prepulse Inhibition/drug effects , Serotonin Receptor Agonists/pharmacology , Spasms, Infantile/drug therapy , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/metabolism , Disease Models, Animal , Disease Progression , Epileptic Syndromes/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Phosphorylation/drug effects , Prepulse Inhibition/physiology , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Random Allocation , Receptors, Serotonin/metabolism , Spasms, Infantile/metabolism
10.
Hum Mol Genet ; 27(12): 2052-2063, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29618004

ABSTRACT

Mutations in the X-linked cyclin-dependent kinase-like 5 (CDKL5) gene cause a complex neurological disorder, characterized by infantile seizures, impairment of cognitive and motor skills and autistic features. Loss of Cdkl5 in mice affects dendritic spine maturation and dynamics but the underlying molecular mechanisms are still far from fully understood. Here we show that Cdkl5 deficiency in primary hippocampal neurons leads to deranged expression of the alpha-amino-3-hydroxy-5-methyl-4-iso-xazole propionic acid receptors (AMPA-R). In particular, a dramatic reduction of expression of the GluA2 subunit occurs concomitantly with its hyper-phosphorylation on Serine 880 and increased ubiquitination. Consequently, Cdkl5 silencing skews the composition of membrane-inserted AMPA-Rs towards the GluA2-lacking calcium-permeable form. Such derangement is likely to contribute, at least in part, to the altered synaptic functions and cognitive impairment linked to loss of Cdkl5. Importantly, we find that tianeptine, a cognitive enhancer and antidepressant drug, known to recruit and stabilise AMPA-Rs at the synaptic sites, can normalise the expression of membrane inserted AMPA-Rs as well as the number of PSD-95 clusters, suggesting its therapeutic potential for patients with mutations in CDKL5.


Subject(s)
Epileptic Syndromes/drug therapy , Protein Serine-Threonine Kinases/genetics , Receptors, AMPA/genetics , Spasms, Infantile/drug therapy , Thiazepines/administration & dosage , Animals , Antidepressive Agents/administration & dosage , Disks Large Homolog 4 Protein/genetics , Epileptic Syndromes/genetics , Epileptic Syndromes/pathology , Gene Expression Regulation/drug effects , Hippocampus/drug effects , Hippocampus/physiopathology , Humans , Mice , Mutation , Neurogenesis/drug effects , Neurons/drug effects , Neurons/pathology , Phosphorylation , Primary Cell Culture , Protein Serine-Threonine Kinases/deficiency , Spasms, Infantile/genetics , Spasms, Infantile/pathology , Synapses/drug effects , Synapses/genetics
11.
Sci Rep ; 7(1): 6228, 2017 07 24.
Article in English | MEDLINE | ID: mdl-28740074

ABSTRACT

The cyclin-dependent kinase-like 5 (CDKL5) gene has been associated with rare neurodevelopmental disorders characterized by the early onset of seizures and intellectual disability. The CDKL5 protein is widely expressed in most tissues and cells with both nuclear and cytoplasmic localization. In post-mitotic neurons CDKL5 is mainly involved in dendritic arborization, axon outgrowth, and spine formation while in proliferating cells its function is still largely unknown. Here, we report that CDKL5 localizes at the centrosome and at the midbody in proliferating cells. Acute inactivation of CDKL5 by RNA interference (RNAi) leads to multipolar spindle formation, cytokinesis failure and centrosome accumulation. At the molecular level, we observed that, among the several midbody components we analyzed, midbodies of CDKL5-depleted cells were devoid of HIPK2 and its cytokinesis target, the extrachromosomal histone H2B phosphorylated at S14. Of relevance, expression of the phosphomimetic mutant H2B-S14D, which is capable of overcoming cytokinesis failure in HIPK2-defective cells, was sufficient to rescue spindle multipolarity in CDKL5-depleted cells. Taken together, these results highlight a hitherto unknown role of CDKL5 in regulating faithful cell division by guaranteeing proper HIPK2/H2B functions at the midbody.


Subject(s)
Carrier Proteins/metabolism , Cell Division , Centrosome/metabolism , Cytokinesis/physiology , Protein Serine-Threonine Kinases/metabolism , Animals , Carrier Proteins/genetics , Cell Cycle , HeLa Cells , Humans , Mice , Phosphorylation , Protein Serine-Threonine Kinases/genetics
12.
Hum Mol Genet ; 26(18): 3520-3530, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28641386

ABSTRACT

CDKL5 is a protein kinase that plays a key role for neuronal functions as testified by the onset of complex neuronal dysfunctions in patients with genetic lesions in CDKL5. Here we identify a novel interactor of CDKL5, IQGAP1, a fundamental regulator of cell migration and polarity. In accordance with a functional role of this interaction, depletion of CDKL5 impairs cell migration and impedes the localization of IQGAP1 at the leading edge. Moreover, we demonstrate that CDKL5 is required for IQGAP1 to form a functional complex with its effectors, Rac1 and the microtubule plus end tracking protein CLIP170. These defects eventually impact on the microtubule association of CLIP170, thus deranging their dynamics. CLIP170 is a cellular target of the neurosteroid pregnenolone; by blocking CLIP170 in its active conformation, pregnenolone is capable of restoring the microtubule association of CLIP170 in CDKL5 deficient cells and rescuing morphological defects in neurons devoid of CDKL5. These findings provide novel insights into CDKL5 functions and pave the way for target-specific therapeutic strategies for individuals affected with CDKL5-disorder.


Subject(s)
Pregnenolone/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Actins/metabolism , Animals , COS Cells , Cell Movement/physiology , Chlorocebus aethiops , HeLa Cells , Humans , Mice , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Microtubules/pathology , Neoplasm Proteins/metabolism , Neurons/metabolism , Neurotransmitter Agents/metabolism , Protein Binding , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism
13.
Sci Rep ; 6: 28295, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27323888

ABSTRACT

MeCP2 is a transcriptional regulator whose functional alterations are responsible for several autism spectrum and mental disorders. Post-translational modifications (PTMs), and particularly differential phosphorylation, modulate MeCP2 function in response to diverse stimuli. Understanding the detailed role of MeCP2 phosphorylation is thus instrumental to ascertain how MeCP2 integrates the environmental signals and directs its adaptive transcriptional responses. The evolutionarily conserved serine 164 (S164) was found phosphorylated in rodent brain but its functional role has remained uncharacterized. We show here that phosphorylation of S164 in brain is dynamically regulated during neuronal maturation. S164 phosphorylation highly impairs MeCP2 binding to DNA in vitro and largely affects its nucleosome binding and chromatin affinity in vivo. Strikingly, the chromatin-binding properties of the global MeCP2 appear also extensively altered during the course of brain maturation. Functional assays reveal that proper temporal regulation of S164 phosphorylation controls the ability of MeCP2 to regulate neuronal morphology. Altogether, our results support the hypothesis of a complex PTM-mediated functional regulation of MeCP2 potentially involving a still poorly characterized epigenetic code. Furthermore, they demonstrate the relevance of the Intervening Domain of MeCP2 for binding to DNA.


Subject(s)
Brain/metabolism , Chromatin/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Animals , DNA Methylation , Dendrites/metabolism , HEK293 Cells , Humans , Mice , Mice, Knockout , NIH 3T3 Cells , Nucleosomes/metabolism , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , Protein Transport , Rabbits , Serine/metabolism
14.
PLoS One ; 11(6): e0157758, 2016.
Article in English | MEDLINE | ID: mdl-27315173

ABSTRACT

Mutations in the X-linked Cyclin-Dependent Kinase-Like 5 gene (CDKL5) cause early onset infantile spasms and subsequent severe developmental delay in affected children. Deleterious mutations have been reported to occur throughout the CDKL5 coding region. Several studies point to a complex CDKL5 gene structure in terms of exon usage and transcript expression. Improvements in molecular diagnosis and more extensive research into the neurobiology of CDKL5 and pathophysiology of CDKL5 disorders necessitate an updated analysis of the gene. In this study, we have analysed human and mouse CDKL5 transcript patterns both bioinformatically and experimentally. We have characterised the predominant brain isoform of CDKL5, a 9.7 kb transcript comprised of 18 exons with a large 6.6 kb 3'-untranslated region (UTR), which we name hCDKL5_1. In addition we describe new exonic regions and a range of novel splice and UTR isoforms. This has enabled the description of an updated gene model in both species and a standardised nomenclature system for CDKL5 transcripts. Profiling revealed tissue- and brain development stage-specific differences in expression between transcript isoforms. These findings provide an essential backdrop for the diagnosis of CDKL5-related disorders, for investigations into the basic biology of this gene and its protein products, and for the rational design of gene-based and molecular therapies for these disorders.


Subject(s)
Alternative Splicing/genetics , Protein Serine-Threonine Kinases/genetics , Spasms, Infantile/genetics , Transcription, Genetic , Amino Acid Sequence , Animals , Exons/genetics , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Humans , Infant, Newborn , Mice , Mutation , Phenotype , Polyadenylation/genetics , Protein Isoforms , Protein Serine-Threonine Kinases/biosynthesis , Spasms, Infantile/pathology
15.
PLoS One ; 11(4): e0153473, 2016.
Article in English | MEDLINE | ID: mdl-27097329

ABSTRACT

MECP2 mutations cause a number of neurological disorders of which Rett syndrome (RTT) represents the most thoroughly analysed condition. Many Mecp2 mouse models have been generated through the years; their validity is demonstrated by the presence of a broad spectrum of phenotypes largely mimicking those manifested by RTT patients. These mouse models, between which the C57BL/6 Mecp2tm1.1Bird strain probably represents the most used, enabled to disclose much of the roles of Mecp2. However, small litters with little viability and poor maternal care hamper the maintenance of the colony, thus limiting research on such animals. For this reason, past studies often used Mecp2 mouse models on mixed genetic backgrounds, thus opening questions on whether modifier genes could be responsible for at least part of the described effects. To verify this possibility, and facilitate the maintenance of the Mecp2 colony, we transferred the Mecp2tm1.1Bird allele on the stronger CD1 background. The CD1 strain is easier to maintain and largely recapitulates the phenotypes already described in Mecp2-null mice. We believe that this mouse model will foster the research on RTT.


Subject(s)
Genetic Background , Methyl-CpG-Binding Protein 2/deficiency , Methyl-CpG-Binding Protein 2/genetics , Models, Animal , Alleles , Animals , Behavior, Animal , Female , Gene Deletion , Male , Mice , Neurons/cytology , Phenotype , Species Specificity
16.
PLoS One ; 11(2): e0148634, 2016.
Article in English | MEDLINE | ID: mdl-26849555

ABSTRACT

In the last years, the X-linked cyclin-dependent kinase-like 5 (CDKL5) gene has been associated with epileptic encephalopathies characterized by the early onset of intractable epilepsy, severe developmental delay, autistic features, and often the development of Rett syndrome-like features. Still, the role of CDKL5 in neuronal functions is not fully understood. By way of a yeast two hybrid screening we identified the interaction of CDKL5 with shootin1, a brain specific protein acting as a determinant of axon formation during neuronal polarization. We found evidence that CDKL5 is involved, at least in part, in regulating neuronal polarization through its interaction with shootin1. Indeed, the two proteins interact in vivo and both are localized in the distal tip of outgrowing axons. By using primary hippocampal neurons as model system we find that adequate CDKL5 levels are required for axon specification. In fact, a significant number of neurons overexpressing CDKL5 is characterized by supernumerary axons, while the silencing of CDKL5 disrupts neuronal polarization. Interestingly, shootin1 phosphorylation is reduced in neurons silenced for CDKL5 suggesting that the kinase affects, directly or indirectly, the post-translational modification of shootin1. Finally, we find that the capacity of CDKL5 to generate surplus axons is attenuated in neurons with reduced shootin1 levels, in agreement with the notion that two proteins act in a common pathway. Altogether, these results point to a role of CDKL5 in the early steps of neuronal differentiation that can be explained, at least in part, by its association with shootin1.


Subject(s)
Axons/metabolism , Cell Polarity/physiology , Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Hippocampus/cytology , Mice , Nerve Tissue Proteins/genetics , Protein Serine-Threonine Kinases/genetics
17.
Cereb Cortex ; 26(6): 2517-2529, 2016 06.
Article in English | MEDLINE | ID: mdl-25979088

ABSTRACT

MeCP2 is associated with several neurological disorders; of which, Rett syndrome undoubtedly represents the most frequent. Its molecular roles, however, are still unclear, and data from animal models often describe adult, symptomatic stages, while MeCP2 functions during embryonic development remain elusive. We describe the pattern and timing of Mecp2 expression in the embryonic neocortex highlighting its low but consistent expression in virtually all cells and show the unexpected occurrence of transcriptional defects in the Mecp2 null samples at a stage largely preceding the onset of overt symptoms. Through the deregulated expression of ionic channels and glutamatergic receptors, the lack of Mecp2 during early neuronal maturation leads to the reduction in the neuronal responsiveness to stimuli. We suggest that such features concur to morphological alterations that begin affecting Mecp2 null neurons around the perinatal age and become evident later in adulthood. We indicate MeCP2 as a key modulator of the transcriptional mechanisms regulating cerebral cortex development. Neurological phenotypes of MECP2 patients could thus be the cumulative result of different adverse events that are already present at stages when no obvious signs of the pathology are evident and are worsened by later impairments affecting the central nervous system during maturation and maintenance of its functionality.


Subject(s)
Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Neurons/metabolism , Animals , Calcium/metabolism , Cell Movement/physiology , Cells, Cultured , Cerebral Cortex/pathology , Disease Progression , Electric Stimulation , Fluorescent Antibody Technique , Glutamic Acid/metabolism , Methyl-CpG-Binding Protein 2/genetics , Mice, Knockout , Neurons/pathology , Rett Syndrome , Time Factors , Transcription, Genetic , Voltage-Sensitive Dye Imaging
18.
PLoS One ; 10(6): e0130183, 2015.
Article in English | MEDLINE | ID: mdl-26098633

ABSTRACT

Rett syndrome (RTT) is an autism spectrum disorder mainly caused by mutations in the X-linked MECP2 gene and affecting roughly 1 out of 10.000 born girls. Symptoms range in severity and include stereotypical movement, lack of spoken language, seizures, ataxia and severe intellectual disability. Notably, muscle tone is generally abnormal in RTT girls and women and the Mecp2-null mouse model constitutively reflects this disease feature. We hypothesized that MeCP2 in muscle might physiologically contribute to its development and/or homeostasis, and conversely its defects in RTT might alter the tissue integrity or function. We show here that a disorganized architecture, with hypotrophic fibres and tissue fibrosis, characterizes skeletal muscles retrieved from Mecp2-null mice. Alterations of the IGF-1/Akt/mTOR pathway accompany the muscle phenotype. A conditional mouse model selectively depleted of Mecp2 in skeletal muscles is characterized by healthy muscles that are morphologically and molecularly indistinguishable from those of wild-type mice raising the possibility that hypotonia in RTT is mainly, if not exclusively, mediated by non-cell autonomous effects. Our results suggest that defects in paracrine/endocrine signaling and, in particular, in the GH/IGF axis appear as the major cause of the observed muscular defects. Remarkably, this is the first study describing the selective deletion of Mecp2 outside the brain. Similar future studies will permit to unambiguously define the direct impact of MeCP2 on tissue dysfunctions.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Insulin-Like Growth Factor I/metabolism , Methyl-CpG-Binding Protein 2/genetics , Muscle Hypotonia/pathology , Muscular Atrophy/pathology , Animals , Disease Models, Animal , Female , Fibrosis/genetics , Fibrosis/pathology , Growth Hormone/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Hypotonia/genetics , Muscle, Skeletal/growth & development , Muscle, Skeletal/pathology , Muscular Atrophy/genetics , Paracrine Communication/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rett Syndrome/genetics , Rett Syndrome/pathology , TOR Serine-Threonine Kinases/metabolism
19.
J Biol Chem ; 290(7): 4512-27, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25555910

ABSTRACT

Mutations in the X-linked CDKL5 (cyclin-dependent kinase-like 5) gene have been associated with several forms of neurodevelopmental disorders, including atypical Rett syndrome, autism spectrum disorders, and early infantile epileptic encephalopathy. Accordingly, loss of CDKL5 in mice results in autistic-like features and impaired neuronal communication. Although the biological functions of CDKL5 remain largely unknown, recent pieces of evidence suggest that CDKL5 is involved in neuronal plasticity. Herein, we show that, at all stages of development, neuronal depolarization induces a rapid increase in CDKL5 levels, mostly mediated by extrasomatic synthesis. In young neurons, this induction is prolonged, whereas in more mature neurons, NMDA receptor stimulation induces a protein phosphatase 1-dependent dephosphorylation of CDKL5 that is mandatory for its proteasome-dependent degradation. As a corollary, neuronal activity leads to a prolonged induction of CDKL5 levels in immature neurons but to a short lasting increase of the kinase in mature neurons. Recent results demonstrate that many genes associated with autism spectrum disorders are crucial components of the activity-dependent signaling networks regulating the composition, shape, and strength of the synapse. Thus, we speculate that CDKL5 deficiency disrupts activity-dependent signaling and the consequent synapse development, maturation, and refinement.


Subject(s)
Neurons/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Phosphatase 1/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/physiology , Animals , Apoptosis , Blotting, Western , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Electrophysiology , Fluorescent Antibody Technique , Hippocampus/cytology , Hippocampus/metabolism , Mice , Neurons/cytology , Phosphorylation , Protein Biosynthesis , Protein Phosphatase 1/genetics , Protein Serine-Threonine Kinases/genetics , Proteolysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, N-Methyl-D-Aspartate/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
20.
J Biol Chem ; 290(6): 3223-37, 2015 Feb 06.
Article in English | MEDLINE | ID: mdl-25527496

ABSTRACT

Mutations in MECP2 cause a broad spectrum of neuropsychiatric disorders of which Rett syndrome represents the best defined condition. Both neuronal and non-neuronal functions of the methyl-binding protein underlie the related pathologies. Nowadays MeCP2 is recognized as a multifunctional protein that modulates its activity depending on its protein partners and posttranslational modifications. However, we are still missing a comprehensive understanding of all MeCP2 functions and their involvement in the related pathologies. The study of human mutations often offers the possibility of clarifying the functions of a protein. Therefore, we decided to characterize a novel MeCP2 phospho-isoform (Tyr-120) whose relevance was suggested by a Rett syndrome patient carrying a Y120D substitution possibly mimicking a constitutively phosphorylated state. Unexpectedly, we found MeCP2 and its Tyr-120 phospho-isoform enriched at the centrosome both in dividing and postmitotic cells. The molecular and functional connection of MeCP2 to the centrosome was further reinforced through cellular and biochemical approaches. We show that, similar to many centrosomal proteins, MeCP2 deficiency causes aberrant spindle geometry, prolonged mitosis, and defects in microtubule nucleation. Collectively, our data indicate a novel function of MeCP2 that might reconcile previous data regarding the role of MeCP2 in cell growth and cytoskeleton stability and that might be relevant to understand some aspects of MeCP2-related conditions. Furthermore, they link the Tyr-120 residue and its phosphorylation to cell division, prompting future studies on the relevance of Tyr-120 for cortical development.


Subject(s)
Centrosome/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , HEK293 Cells , HeLa Cells , Humans , Methyl-CpG-Binding Protein 2/genetics , Mice , Microtubules/metabolism , Mitosis , Mutation, Missense , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport , Rett Syndrome/genetics
SELECTION OF CITATIONS
SEARCH DETAIL